Cargando…

Anti-cancer potency by induced apoptosis by molecular docking P53, caspase, cyclin D1, cytotoxicity analysis and phagocytosis activity of trisindoline 1,3 and 4

Cancer is one of the leading causes of death in the world. Efforts to find and develop cancer drugs from natural products continue with the exploration of trisindoline, a substance that is isolated from marine sponges Hyrtios altum. Trisindoline is an indole trimer alkaloid compound that has been su...

Descripción completa

Detalles Bibliográficos
Autores principales: Nurhayati, Awik Puji Dyah, Rihandoko, Andis, Fadlan, Arif, Ghaissani, Shabrina Syifa, Jadid, Nurul, Setiawan, Edwin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Elsevier 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9563049/
https://www.ncbi.nlm.nih.gov/pubmed/36249936
http://dx.doi.org/10.1016/j.jsps.2022.06.012
_version_ 1784808315066253312
author Nurhayati, Awik Puji Dyah
Rihandoko, Andis
Fadlan, Arif
Ghaissani, Shabrina Syifa
Jadid, Nurul
Setiawan, Edwin
author_facet Nurhayati, Awik Puji Dyah
Rihandoko, Andis
Fadlan, Arif
Ghaissani, Shabrina Syifa
Jadid, Nurul
Setiawan, Edwin
author_sort Nurhayati, Awik Puji Dyah
collection PubMed
description Cancer is one of the leading causes of death in the world. Efforts to find and develop cancer drugs from natural products continue with the exploration of trisindoline, a substance that is isolated from marine sponges Hyrtios altum. Trisindoline is an indole trimer alkaloid compound that has been successfully synthesized into trisindoline 1, 3 and 4. Trisindoline is cytotoxic in cell lines and in this study, trisindoline was able to induce apoptosis in the in silico and in vitro tests that were carried out. The in silico test was carried out through molecular docking using the Autodock Vina method and the Molecular Dynamics (MD) Simulation QM / MM AMBER. The target proteins used were protein p53 and caspase −9 which played a role in the apoptotic pathway and cyclin D1 which played a role in cell proliferation. Meanwhile, cytotoxicity analysis was carried out using the MTT method (3- (4,5-dimethyltiazol −2-yl) −2,5 -dipenyl tetrazolium bromide). Nevertheless, the ability of trisindoline to induce phagocytosis is still unrevealed. The phagocytosis assay was carried out by assessing the macrophage capacity and phagocytic index using the latex-beads model. The in silico results showed that the binding affinity values between the target protein Cdk-2 and the trisindoline 1, trisindoline 3 and trisindoline 4 ligands were −7.3 kcal / mol, −7.7 kcal / mol and −6.6 kcal / mol respectively. The binding affinity values between the target protein p53 and the trisindoline 1, trisindoline 3 and trisindoline 4 ligands were −7.5 kcal / mol, −7.4 kcal / mol and −7.5 kcal / mol respectively. The binding affinity values between the target protein caspase-9 and the trisindoline 1, trisindoline 3 and trisindoline 4 ligands were −7.5 kcal / mol, −7.1 kcal / mol and −7.2 kcal / mol respectively. The results of RMSD (Root Mean Square Deviation), RMSF (Root Mean Square Fluctuation), and hydrogen bonds in the MD (Molecular Dynamics) Simulation showed that Cdk-2 formed a protein complex with trisindoline 3, protein p53 with trisindoline 1 and caspase-9 with trisindoline 1. The cytotoxicity assay was carried out in the MCF-7 cell line and the IC50 value obtained for trisindoline 1 was 2.059 μM, for trisindoline 3 was 3.9759 ​​μM, for trisindoline 4 was 15.46 μM and for doxorubicin was 9.88 μM. Furthermore, the phagocytosis test was carried out using trisindoline 1, 3 and 4. Our results showed that 6.25 µg mL(−1) of trisindoline 1 and trisindoline 3 were able to induce the phagocytosis capacity of macrophage cells of 38.34; whereas trisindoline 4 at a concentration of 50 µg mL(−1) induces a phagocytosis capacity of 32.89. Trisindoline 1, 3 and 4 showed potentials of immunostimulants at low concentrations but showed potentials of immunosuppressants at high concentrations. The overall results demonstrated that trisindoline 1 and 3 are potential anti-cancer candidates capable of activating the apoptotic pathway.
format Online
Article
Text
id pubmed-9563049
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Elsevier
record_format MEDLINE/PubMed
spelling pubmed-95630492022-10-15 Anti-cancer potency by induced apoptosis by molecular docking P53, caspase, cyclin D1, cytotoxicity analysis and phagocytosis activity of trisindoline 1,3 and 4 Nurhayati, Awik Puji Dyah Rihandoko, Andis Fadlan, Arif Ghaissani, Shabrina Syifa Jadid, Nurul Setiawan, Edwin Saudi Pharm J Original Article Cancer is one of the leading causes of death in the world. Efforts to find and develop cancer drugs from natural products continue with the exploration of trisindoline, a substance that is isolated from marine sponges Hyrtios altum. Trisindoline is an indole trimer alkaloid compound that has been successfully synthesized into trisindoline 1, 3 and 4. Trisindoline is cytotoxic in cell lines and in this study, trisindoline was able to induce apoptosis in the in silico and in vitro tests that were carried out. The in silico test was carried out through molecular docking using the Autodock Vina method and the Molecular Dynamics (MD) Simulation QM / MM AMBER. The target proteins used were protein p53 and caspase −9 which played a role in the apoptotic pathway and cyclin D1 which played a role in cell proliferation. Meanwhile, cytotoxicity analysis was carried out using the MTT method (3- (4,5-dimethyltiazol −2-yl) −2,5 -dipenyl tetrazolium bromide). Nevertheless, the ability of trisindoline to induce phagocytosis is still unrevealed. The phagocytosis assay was carried out by assessing the macrophage capacity and phagocytic index using the latex-beads model. The in silico results showed that the binding affinity values between the target protein Cdk-2 and the trisindoline 1, trisindoline 3 and trisindoline 4 ligands were −7.3 kcal / mol, −7.7 kcal / mol and −6.6 kcal / mol respectively. The binding affinity values between the target protein p53 and the trisindoline 1, trisindoline 3 and trisindoline 4 ligands were −7.5 kcal / mol, −7.4 kcal / mol and −7.5 kcal / mol respectively. The binding affinity values between the target protein caspase-9 and the trisindoline 1, trisindoline 3 and trisindoline 4 ligands were −7.5 kcal / mol, −7.1 kcal / mol and −7.2 kcal / mol respectively. The results of RMSD (Root Mean Square Deviation), RMSF (Root Mean Square Fluctuation), and hydrogen bonds in the MD (Molecular Dynamics) Simulation showed that Cdk-2 formed a protein complex with trisindoline 3, protein p53 with trisindoline 1 and caspase-9 with trisindoline 1. The cytotoxicity assay was carried out in the MCF-7 cell line and the IC50 value obtained for trisindoline 1 was 2.059 μM, for trisindoline 3 was 3.9759 ​​μM, for trisindoline 4 was 15.46 μM and for doxorubicin was 9.88 μM. Furthermore, the phagocytosis test was carried out using trisindoline 1, 3 and 4. Our results showed that 6.25 µg mL(−1) of trisindoline 1 and trisindoline 3 were able to induce the phagocytosis capacity of macrophage cells of 38.34; whereas trisindoline 4 at a concentration of 50 µg mL(−1) induces a phagocytosis capacity of 32.89. Trisindoline 1, 3 and 4 showed potentials of immunostimulants at low concentrations but showed potentials of immunosuppressants at high concentrations. The overall results demonstrated that trisindoline 1 and 3 are potential anti-cancer candidates capable of activating the apoptotic pathway. Elsevier 2022-09 2022-06-21 /pmc/articles/PMC9563049/ /pubmed/36249936 http://dx.doi.org/10.1016/j.jsps.2022.06.012 Text en © 2022 The Authors https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Original Article
Nurhayati, Awik Puji Dyah
Rihandoko, Andis
Fadlan, Arif
Ghaissani, Shabrina Syifa
Jadid, Nurul
Setiawan, Edwin
Anti-cancer potency by induced apoptosis by molecular docking P53, caspase, cyclin D1, cytotoxicity analysis and phagocytosis activity of trisindoline 1,3 and 4
title Anti-cancer potency by induced apoptosis by molecular docking P53, caspase, cyclin D1, cytotoxicity analysis and phagocytosis activity of trisindoline 1,3 and 4
title_full Anti-cancer potency by induced apoptosis by molecular docking P53, caspase, cyclin D1, cytotoxicity analysis and phagocytosis activity of trisindoline 1,3 and 4
title_fullStr Anti-cancer potency by induced apoptosis by molecular docking P53, caspase, cyclin D1, cytotoxicity analysis and phagocytosis activity of trisindoline 1,3 and 4
title_full_unstemmed Anti-cancer potency by induced apoptosis by molecular docking P53, caspase, cyclin D1, cytotoxicity analysis and phagocytosis activity of trisindoline 1,3 and 4
title_short Anti-cancer potency by induced apoptosis by molecular docking P53, caspase, cyclin D1, cytotoxicity analysis and phagocytosis activity of trisindoline 1,3 and 4
title_sort anti-cancer potency by induced apoptosis by molecular docking p53, caspase, cyclin d1, cytotoxicity analysis and phagocytosis activity of trisindoline 1,3 and 4
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9563049/
https://www.ncbi.nlm.nih.gov/pubmed/36249936
http://dx.doi.org/10.1016/j.jsps.2022.06.012
work_keys_str_mv AT nurhayatiawikpujidyah anticancerpotencybyinducedapoptosisbymoleculardockingp53caspasecyclind1cytotoxicityanalysisandphagocytosisactivityoftrisindoline13and4
AT rihandokoandis anticancerpotencybyinducedapoptosisbymoleculardockingp53caspasecyclind1cytotoxicityanalysisandphagocytosisactivityoftrisindoline13and4
AT fadlanarif anticancerpotencybyinducedapoptosisbymoleculardockingp53caspasecyclind1cytotoxicityanalysisandphagocytosisactivityoftrisindoline13and4
AT ghaissanishabrinasyifa anticancerpotencybyinducedapoptosisbymoleculardockingp53caspasecyclind1cytotoxicityanalysisandphagocytosisactivityoftrisindoline13and4
AT jadidnurul anticancerpotencybyinducedapoptosisbymoleculardockingp53caspasecyclind1cytotoxicityanalysisandphagocytosisactivityoftrisindoline13and4
AT setiawanedwin anticancerpotencybyinducedapoptosisbymoleculardockingp53caspasecyclind1cytotoxicityanalysisandphagocytosisactivityoftrisindoline13and4